Your activity: 24 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Prognosis of acute myeloid leukemia

Prognosis of acute myeloid leukemia
Author:
Charles A Schiffer, MD
Section Editor:
Richard A Larson, MD
Deputy Editor:
Alan G Rosmarin, MD
Literature review current through: Dec 2022. | This topic last updated: Oct 29, 2021.

INTRODUCTION — The term acute myeloid leukemia (AML) refers to a group of hematopoietic neoplasms involving cells committed to the myeloid line of cellular development. AML is characterized by a clonal proliferation of myeloid precursors with reduced capacity to differentiate into more mature cellular elements.

The response to treatment and overall survival of patients with AML is heterogeneous. A number of prognostic factors related to patient and tumor characteristics have been described for AML, including age, performance status, and karyotype (table 1) [1-3].

This topic will review prognostic factors in AML. Adverse risk factors that are more common in older adults (eg, patients over age 60 years) with AML are discussed separately. (See "Pretreatment evaluation and prognosis of acute myeloid leukemia in older adults", section on 'Prognosis'.)

The diagnosis, treatment, complications of AML, and further information on specific cytogenetic abnormalities are also discussed separately. (See "Clinical manifestations, pathologic features, and diagnosis of acute myeloid leukemia" and "Cytogenetic abnormalities in acute myeloid leukemia" and "Induction therapy for acute myeloid leukemia in medically-fit adults" and "Acute myeloid leukemia: Management of medically-unfit adults" and "Overview of the complications of acute myeloid leukemia".)

CLINICAL RISK FACTORS — There are several clinical findings that may help predict the likelihood of attaining a complete remission (CR) and subsequent disease-free survival (DFS) in patients with AML. The strongest adverse clinical predictors are:

Advanced age

Poor performance status

Cytogenetic and/or molecular genetic findings in tumor cells

History of prior exposure to cytotoxic agents or radiation therapy

History of prior myelodysplasia or other hematologic disorders such as myeloproliferative neoplasms

The first two factors are the main predictors of early death, while the others are better predictors of resistant disease or early relapse. (See "Therapy-related myeloid neoplasms: Epidemiology, causes, evaluation, and diagnosis" and 'Karyotype' below.)

Age — While there is no clearly accepted definition of younger compared to older adults when dealing with AML, in most studies, "older adults" was defined as greater than ages 55, 60, or 65 years. Such older adults have lower rates of achieving a CR and shorter DFS when compared with younger patients.

Higher age appears to act as an adverse prognostic marker even in younger patients. A population-based retrospective study from the United Kingdom that included 11,303 patients with AML diagnosed from 2001 to 2006 reported an estimated five-year overall survival (OS) rate of 15 percent, which varied according to the age at diagnosis [4]:

15 to 24 years (289 patients) – 53 percent

25 to 39 years (702 patients) – 49 percent

40 to 59 years (2170 patients) – 33 percent

60 to 69 years (2208 patients) – 13 percent

70 to 79 years (3258 patients) – 3 percent

>80 years (2676 patients) – 0 percent

A retrospective analysis of 891 patients <30 years old found that five-year event-free survival (EFS) rates decreased as patient age at diagnosis increased with rates of 54, 46, and 28 percent for children (2 to <13 years), adolescents (13 to <21 years), and young adults (21 to <30 years), respectively [5]. With favorable karyotypes excluded, outcomes were superior in children (EFS 44 percent) compared with adolescents (35 percent) and young adults (23 percent).

The effect of age on outcome in patients with AML is discussed in more detail separately. (See "Pretreatment evaluation and prognosis of acute myeloid leukemia in older adults", section on 'Prognosis'.)

Performance status — Most other clinical risk factors in AML are related to comorbidities (eg, heart failure, renal insufficiency, concurrent infection) that can be partially reflected by the patient's performance status [3,6]. The two most commonly used tools are the Karnofsky performance status and the Eastern Cooperative Oncology Group (ECOG) performance status (table 2A-B). Performance status and age at diagnosis can be combined to estimate the percentage of patients who will die within the first 28 days of treatment [3]. This ranges from 5 percent for patients under the age of 50 years with an ECOG performance status <3 to 57 percent for patients over age 69 with an ECOG performance status of ≥3 (table 3). Performance status appears to be of greatest prognostic value in older adults and may not predict early outcomes (mortality, intensive care unit admission, CR) in younger patients [7]. (See "Pretreatment evaluation and prognosis of acute myeloid leukemia in older adults", section on 'Physical functioning'.)

As an example, a retrospective analysis of 968 adults with newly diagnosed AML treated by the Southwest Oncology Group (SWOG) enrolled in prospective trials of induction therapy reported increasing 30-day mortality rate with worsening performance status and increased age at diagnosis [8]. Patients ≤65 years of age had 30-day mortality rates of 5, 4, 9, and 21 percent if they presented with an ECOG performance status of 0, 1, 2, or 3, respectively. Patients >65 years had rates of 13, 16, 35, and 60 percent, respectively. Older patients with a worse performance status were also less likely to obtain a CR. However, subsequent studies have reported lower rates of short-term mortality in older patients, probably due to improvements in supportive care. As an example, a large randomized trial in patients >60 years of age reported a 30-day mortality of approximately 11 percent [9].

It should be emphasized that these and other published estimates of outcomes derive from results of patients entered on clinical trials. These trials usually exclude individuals with organ dysfunction and substantially impaired performance status and hence these results may not directly apply to the large fraction of older patients with medical and sometimes psychosocial comorbidities. Indeed, data indicate that the majority of older patients do not receive induction chemotherapy as a consequence of either their refusal of treatment or physician assessment that they would not tolerate or benefit from treatment. Assessment of performance status in AML can be tricky. For example, a patient may have no or minimal symptoms with a PS of 0 or 1 and develop fever and infection and instantly become a PS 3 or 4, depending on the severity of the infection. With proper treatment, they can rapidly revert to baseline. Therefore, many factors may have to be considered when using PS to predict prognosis, and it may be that medical comorbidity indices that also include more chronic medical conditions will be more helpful in the future. A number of such scoring systems, some originally designed for transplant recipients, have been developed, but none have been validated for use in individual patients.

Therapy-related AML — Persons who are exposed to cytotoxic agents or radiation therapy are at risk of developing AML, myelodysplastic syndrome (MDS), and myelodysplastic syndrome/myeloproliferative neoplasms (MDS/MPN). These conditions lie along a continuum of disease and are categorized by the 2016 World Health Organization classification system as therapy-related myeloid neoplasms (t-MN) [10]. T-MNs account for approximately 10 to 20 percent of all cases of AML, MDS, and MDS/MPN. The incidence among patients treated with cytotoxic agents varies according to the underlying disease, specific agents, timing of exposure, and dose. The prognosis of patients with t-MN is generally worse than for those with de novo AML because of higher rates of drug resistance. This is discussed in more detail separately. (See "Therapy-related myeloid neoplasms: Epidemiology, causes, evaluation, and diagnosis" and "Cytogenetic abnormalities in acute myeloid leukemia", section on 'Therapy-related myeloid neoplasms'.)

Antecedent hematologic disorders — Pre-existing myelodysplastic or myeloproliferative disorders are common in older patients with AML, occurring in 24 to 40 percent of cases [11-15]. These disorders are often associated with ineffective hematopoiesis and dysfunctional blood cells. By the time that AML emerges, these patients may be colonized by pathogenic flora, threatened by recurrent bleeding episodes, and dependent on transfusions.

In one study that compared gene mutations in patients with secondary AML (after a prior myeloid malignancy), therapy-related AML, and unspecified AML, the identification of a gene mutation in one of eight genes (SRSF2, SF3B1, U2AF1, ZRSR2, ASXL1, EZH2, BCOR, or STAG2) was highly specific for a diagnosis of secondary AML and a poor clinical outcome [16].

Other factors — The effect of race on patient outcomes in AML is uncertain. Using data from seven CALGB studies, it was found that African American and White subjects with AML differ significantly with respect to important risk factors such as cytogenetic risk groups and age [1]. African-American men had significantly lower rates of CR (54 versus 64 percent) and five-year OS (16 versus 24 percent) than White men, and may represent a higher risk group. Further studies of this type are needed to confirm these results, suggest possible biologic mechanisms, and provide appropriate high-risk treatment programs.

CYTOGENETIC AND MOLECULAR FEATURES — Specific cytogenetic and molecular features permit stratification of AML into various prognostic groups. Understanding of the impact of combinations of cytogenetic and molecular findings is evolving. We prefer an approach similar to the classification system proposed by the European LeukemiaNet (ELN) (table 4) [17]. Treatment decisions that are informed by risk stratification are discussed separately. (See "Post-remission therapy for acute myeloid leukemia in younger adults" and "Treatment of relapsed or refractory acute myeloid leukemia".)

Information regarding the genetic landscape of AML and its impact on prognosis comes from an analysis of 1540 patients with AML enrolled on prospective trials of intensive therapy that included correlative studies with cytogenetic analysis and gene sequencing [18]. This study identified 14 major genomic subgroups of AML that largely correspond to the ELN classification.

European LeukemiaNet classification — The ELN integrates cytogenetic and molecular features (eg, FLT3-ITD, CEBPA, and NPM1) in AML to divide cases into three prognostic risk groups that differ based on rates of complete remission (CR), disease-free survival (DFS), and overall survival (OS) (table 4) [17]:

Favorable risk:

t(8;21)(q22;q22.1); RUNX1-RUNX1T1

inv(16)(p13.1;q22) or t(16;16)(p13.1;q22); CBFB-MYH11

Mutated NPM1 without FLT3-ITD or with low allelic ratio (<0.5) of FLT3-ITD

Biallelic mutated CEBPA

Intermediate risk:

Mutated NPM1 and high allelic ratio (>0.5) of FLT3-ITD

Wild-type NPM1 without FLT3-ITD or with low allelic ratio (<0.5) of FLT3-ITD (without adverse-risk genetic lesions)

t(9;11)(p21.3;q23.3); MLLT3-KMT2A

Cytogenetic abnormalities not classified as favorable or adverse

Adverse risk:

t(6;9)(p23;q34.1); DEK-NUP214

t(v;11q23.3); KMT2A rearranged

t(9;22)(q34.1;q11.2); BCR-ABL1

inv(3)(q21.3;q26.2) or t(3;3)(q21.3;q26.2); GATA2, MECOM (EVI1)

Monosomy 5 or del(5q); monosomy 7; monosomy 17/abn(17p)

Complex karyotype, monosomal karyotype

Wild-type NPM1 and high allelic ratio (>0.5) of FLT3-ITD

Mutant RUNX1, ASXL1, or TP53

Note the use of "allelic ratio" to stratify based on the FLT3-ITD mutation. Various research groups calculate this ratio differently and/or use other cutoff values. This calculation is affected by the admixture of remaining normal bone marrow cells, which add normal alleles to the denominator, thereby "diluting" or reducing the calculated ratio. Thus, this measure may only be reliable if the majority of the cells are leukemic and the fraction is between 0.5 and 1.0, which would necessarily indicate that the malignant cells are either homozygous or hemizygous for the FLT3 mutant allele.

A "monosomal karyotype" is defined as at least two autosomal monosomies or a single autosomal monosomy in the presence of one or more structural cytogenetic abnormalities. Additional details regarding the stratification scheme are provided in the table (table 4) [17].

Other models include clinical features in addition to cytogenetic and molecular features. The prognostic index for cytogenetically normal AML (PINA) combines cytogenetic and molecular data with patient age, performance status, and white blood cell count at diagnosis to identify three distinct cohorts (low, intermediate, and high risk) with statistically different estimated rates of OS (74, 28, and 3 percent) and cumulative incidence of relapse (35, 56, and 72 percent) at five years [19].

Karyotype

General — Karyotype analysis with metaphase cytogenetics is a key component of the initial evaluation of a patient with AML; specific cytogenetic abnormalities in AML have considerable prognostic significance and affect treatment planning (table 5 and table 4). The value of risk stratification by karyotype has been illustrated in several analyses of patients enrolled in prospective clinical trials. The largest studies were cooperative group efforts from the Medical Research Council (MRC), the Southwest Oncology Group/Eastern Cooperative Oncology Group (SWOG/ECOG), and the Cancer and Leukemia Group B (CALGB). All studies confirmed earlier results from other groups attesting to the importance of pretreatment karyotype (figure 1) [20-27]. The following describes methods of grouping karyotype abnormalities by risk of progression. Details regarding specific chromosomal changes and their impact on prognosis are presented separately. (See "Cytogenetic abnormalities in acute myeloid leukemia".)

The specifics regarding what constitutes favorable, intermediate, and unfavorable risk have varied among the cooperative groups. While there has been general agreement that t(8;21), inv(16), and t(15;17) predict a good outcome, there has been disagreement regarding what abnormalities determine an unfavorable risk and how additional chromosomal abnormalities impact the prognostic value of known markers.

Information from 5876 adults with newly diagnosed de novo (93 percent) or secondary AML enrolled on prospective MRC trials of intensive anthracycline- and cytarabine-based combination chemotherapy [27] was used to modify the MRC's prior stratification system [28,29] to create the risk stratification system shown below. Using these definitions, rates of OS at 10 years were 69, 38, 33, and 12 percent for patients with favorable risk, normal karyotype, intermediate risk, and adverse risk, respectively.

Favorable (approximately 16 percent of newly diagnosed patients) – The following abnormalities are considered favorable, whether alone or in conjunction with other abnormalities: t(8;21), inv(16)(p13;q22), t(16;16)(p13;q22). t(15;17)(q24.1;q21.1) which identifies acute promyelocytic leukemia (APL) is also favorable, but APL is now considered separately from other forms of AML because different treatment approaches are used.

Normal karyotype (approximately 40 percent)

Intermediate (approximately 20 percent) – Abnormalities not described in favorable or unfavorable.

Adverse or poor (approximately 25 percent) – The following abnormalities are considered unfavorable when they occur in cases that do not also contain favorable karyotypic changes: del (5q); add (5q); del (7q); add (7q); monosomies 5 or 7; inv(3)(q21q26); t(3;3)(q21;q26); t(6;11)(q27;q23); t(10;11)(p11-13;q23); t(9;22)(q34;q11); 17p abnormalities or monosomy 17; complex aberrant karyotypes described as at least 4 unrelated abnormalities; 11q23 abnormalities excluding t(9;11)(p21;q23) and excluding t(11;19)(q23;p13); or abnormalities of 3q excluding t(3;5)(q21-25;q31-35).

These survival rates are consistent with those seen by other cooperative groups using similar risk stratification systems [22,30]. This study also served to clarify the prognostic value of findings that were previously not well understood. As examples:

A "monosomal karyotype," defined as at least two autosomal monosomies or a single autosomal monosomy in the presence of one or more structural cytogenetic abnormalities, has been proposed as a better predictor of unfavorable-risk disease than a complex karyotype [31]. The approximately 10 percent of patients with AML who demonstrate a monosomal karyotype have a low rate of CR after induction (48 percent) and a <5 percent rate of OS at four years [31,32]. The additional prognostic value of including "monosomal karyotype" in the adverse risk group was specifically addressed in the analysis of 5876 patients on MRC trials [27]. While patients with a monosomal karyotype had particularly poor outcomes (<5 percent OS at 10 years), 94 percent of cases with a monosomal karyotype were already classified as adverse risk according to the criteria defined above. In at least one retrospective study, the prognostic values of the monosomal karyotype and complex karyotype did not remain when patients with 17p abnormalities or chromosome 5 abnormalities were removed from the analysis [33].

In two large retrospective analyses, patients with a monosomal karyotype had lower rates of CR and OS when compared with patients with unfavorable cytogenetics without a monosomal karyotype [34,35]. The overall outcome for patients with a monosomal karyotype appeared to be better following allogeneic hematopoietic cell transplantation (HCT) when compared with consolidation with chemotherapy alone. The benefit of allogeneic HCT in this population was further supported by an analysis of 305 patients with newly diagnosed AML with a monosomal karyotype enrolled in three consecutive HOVON/SAKK phase III trials [36]. CR was achieved in 140 (46 percent) and 107 proceeded with consolidation chemotherapy (48 patients), high dose chemotherapy with autologous stem cell rescue (14 patients), or allogeneic HCT (45 patients). The rates of overall and relapse-free survival (RFS) at five years were 13 and 12 percent, respectively. The five-year OS rates were higher with allogeneic HCT when compared with chemotherapy or autologous stem cell rescue (19 versus 9 percent). None of the 33 patients who achieved a CR but did not proceed to consolidation survived.

Patients with abnormalities of chromosome band 11q23, seen in 5 to 10 percent of patients with de novo AML and in some patients with therapy-related leukemia after having received topoisomerase II inhibitors, have had a poor outcome when treated with conventional chemotherapy and have previously been included in the adverse risk group [37-40]. Data from the MRC analysis supported prior suggestions that a subset of patients with t(9;11) may be cured with intensive consolidation chemotherapy alone (10 year OS rate of 39 percent) [27,41,42]. As such, patients with this particular 11q23 translocation are not considered to have adverse risk based on this abnormality.

Prior to the MRC analysis, it was not clear how to categorize patients with isolated trisomy 8 with some studies suggesting that these patients had adverse outcomes [43] and others reporting that their outcomes did not differ from those with normal cytogenetics [44]. The 547 patients with trisomy 8 included in the MRC database had a similar prognosis to those with normal cytogenetics (10-year OS in 37 percent) [27].

While patients with cytogenetically normal AML have traditionally been included in the intermediate risk group category, more sophisticated analyses, including gene mutation and microRNA expression studies, suggest that this group is more heterogeneous than previously thought. The effect of gene mutations and microRNA expression on prognosis is discussed in more detail below. (See 'Gene mutations' below and 'MicroRNA expression profiling' below.)

Older adults — Similar stratification systems to that used in the general population (described above) have also shown prognostic value in older adults with AML [11,13,45-48]. The more favorable translocations seen with core-binding factor AML are considerably less common in older adults. In this population, patients with favorable (7 percent), intermediate (73 percent), or poor risk (20 percent) cytogenetics had CR rates of 72, 53 to 63, and 26 percent, respectively [45]. OS rates at five years for the same groups were 34, 10 to 15, and 2 percent, respectively.

A study from the CALGB included 42 patients with isolated trisomy 8, 60 percent of whom were more than 60 years of age [43]. Median survival was lower in trisomy 8 patients over the age of 60 than in younger patients (4.8 versus 17.5 months) with no long-term survivors among the older patients. The only long-term survivors were those less than 60 years of age who were treated with autologous or allogeneic HCT while in first CR (CR1). A later CALGB study indicated a five-year OS of 7 to 9 percent for the group of patients >60 years of age with trisomy 8 [49].

In another CALGB study in patients ≥60 years of age with AML, the presence of a complex karyotype consisting of three or more chromosome abnormalities in at least one clone or a rare aberration (in the absence of t(8;21) and inv(16)), was associated with five-year DFS and OS rates of 2 percent or less following standard cytotoxic chemotherapy used in conventional doses [49]. Thus, such patients are better suited for investigational therapy on a clinical trial or supportive care. Similar conclusions were made in a German-Austrian study, in which three-year OS for patients >70 years of age with high-risk cytogenetics was 2 percent [47].

Gene mutations — AML is typically categorized based on combinations of mutations and/or altered expression of certain genes (table 4). Many centers routinely perform genetic profiling on all newly diagnosed patients. Abnormalities in certain genes (eg, mutations in FLT3, NPM1, KIT) and altered gene expression profiles confer prognostic significance in adult patients with AML [50]. This is particularly important in the approximately 45 percent of patients with normal karyotypes. This heterogeneous group of patients contains some with a better prognosis (eg, mutant CEBPA) and others with an adverse prognosis (eg, FLT3-ITD) (table 1 and table 6) [51-67].

Abnormalities in FLT3, NPM1, KIT, CEBPA, TP53, RUNX1, and ASXL1 have been the most widely studied. Other gene mutations, such as those involving WT1 (Wilms tumor 1), meningioma 1 (MN1), TET2, IDH1, IDH2, DNMT3A, SRSF2, or RAS may also have prognostic significance but need further confirmation in prospective studies [68-92]. (See "Molecular genetics of acute myeloid leukemia", section on 'Mutations affecting DNA methylation'.)

As an example, one report described a panel of 17 genes that reflected the functional leukemic stem cell phenotype of specimens from newly diagnosed AML [93]. The clinical impact of many of the mutations identified by these tests is uncertain, and only a few of these abnormalities are currently "druggable" or targeted by available agents. Hopefully, better understanding of the mechanisms by which these changes produce increased or decreased sensitivity to treatment will evolve, permitting more rational choices of chemotherapy and the selective application of transplantation.

FLT3 gene — FLT3 is a transmembrane tyrosine kinase receptor that stimulates cell proliferation upon activation. Mutations in the FMS-like tyrosine kinase 3 gene producing internal tandem duplications (FLT3-ITD) and constitutive activation of the FLT3 receptor tyrosine kinase are quite common in AML, particularly in patients with normal karyotypes, and have been associated with poorer OS in children and in younger and older adults receiving intensive chemotherapy [56,82,94-105]. It has been proposed that FLT3-ITD mutational status is the primary predictor of outcome among patients with intermediate-risk AML by karyotype analysis [104]. The exact frequency varies with age with FLT3 mutations being present in approximately 10 and 30 percent of patients with cytogenetically normal AML in the pediatric and adult populations, respectively [100,106,107]. Concurrent abnormalities in other genes, such as NPM1, may influence the impact of the FLT3 mutation [108].

There are two main types of FLT3 mutations. The most common are internal tandem duplications (ITD) of different length that result in ligand-independent activation of the FLT3 receptor and a proliferative signal. Additionally, in 5 to 10 percent of patients, point mutations in the activating loop of the kinase domain of FLT3 may also result in activation of FLT3 [109,110]. The prognostic impact of FLT3-ITD is influenced by its mutational context, including the absence of the wild-type FLT3 allele (ie, homozygous or hemizygous FLT3-ITD), the concurrent mutation status of NPM1, and the FLT3 mutant/wild-type allelic ratio (AR). Homozygous or hemizygous FLT3 and higher AR are associated with poorer outcomes [96,111,112]. Patients with mutant NPM1 and FLT3-ITD with a low AR (<0.5) have more favorable outcomes, whereas patients with wild-type NPM1 and FLT3 AR ≥0.5 have poor outcomes [101,113,114].

The MRC conducted an analysis of the results of both autologous and allogeneic HCT in patients whose leukemia cells were positive for FLT3-ITD [115]. This was a retrospective evaluation of large studies of patients who had been randomly assigned to these HCT approaches irrespective of their FLT3 mutational status. In FLT3-ITD positive patients, there was no difference in RFS or OS between patients allocated to either type of HCT and those treated with chemotherapy alone. However, somewhat conflicting results were seen in similar retrospective analyses conducted by German leukemia groups who noted improved outcomes in FLT3 mutated patients who had a histocompatible sibling donor compared with those who did not [101].

In another study, the European Group for Blood and Marrow Transplantation (EBMT) analyzed the outcomes of 206 patients with AML who underwent HLA-identical sibling or matched unrelated allogeneic HCT in CR1 after myeloablative conditioning [116]. The 120 patients with FLT3-ITD had a higher median leukocyte count at diagnosis and a shorter interval from CR to HCT, but otherwise had similar baseline characteristics to those without FLT3-ITD. The presence of FLT3-ITD was associated with a higher estimated cumulative incidence of relapse at two years post-transplant (30 versus 16 percent), and a lower two-year leukemia-free survival rate (58 versus 71 percent).

Use of midostaurin (FLT3 inhibitor) as a component of remission induction therapy and the importance of FLT3 status on selection of post-remission therapy for AML are discussed separately. (See "Induction therapy for acute myeloid leukemia in medically-fit adults", section on 'Inhibitors of mutated FLT3' and "Post-remission therapy for acute myeloid leukemia in younger adults", section on 'Unfavorable-risk disease'.)

NPM gene — Abnormalities in the nucleophosmin (NPM1) gene are found in approximately 25 and 50 percent of patients with de novo AML or de novo normal karyotype AML, respectively. NPM1 mutations have been associated with improved outcomes in younger and older adults, and children with AML, although the mechanism for increased chemosensitivity is not known [1,82,117-121]. However, concurrent cytogenetic abnormalities, mutations in other genes (eg, FLT3), and the NPM1 mutant allele burden influence the impact of the NPM1 mutation [104,122-124]. The superior prognosis is limited to those with NPM1 mutation who do not have a FLT3-ITD mutation and a normal karyotype.

In 215 younger adults with newly diagnosed AML enrolled on prospective MRC trials, patients with a normal karyotype AML and FLT3-ITD with wild-type NPM1 had a poor prognosis (13 percent alive at 10 years) while patients with NPM1 mutation without FLT3-ITD demonstrated superior OS rates (50 percent alive at 10 years) [27]. There were not enough data regarding CEBPA status to analyze its effect on prognosis [101,125-128].

Subjects with a normal karyotype, NPM1 mutations, wild-type FLT3, and low levels of ERG appear to have an especially favorable prognosis, with an estimated two-year PFS of 70 percent after induction treatment with cytarabine, daunorubicin, and etoposide followed by intensification with high dose cytarabine or intensification followed by autologous HCT [56]. In another study, the four-year OS in similar patients was approximately 60 percent [101].

Pooled analysis from nine international studies identified 2426 patients with NPM1 mutations and FLT3-wild type or FLT3-ITD with low allelic burden, including 2000 with normal karyotype and 426 with abnormal karyotype [124]. In patients with this molecular profile, those with adverse cytogenetics had lower rates of CR and five-year OS and event-free survival (EFS), indicating that higher cytogenetic risk negates the favorable molecular risk of NPM1 mutations and FLT3-wild type or FLT3-ITD with low allelic burden. It should be noted that only 3 percent of the overall NPM1 mutated group had a high-risk karyotype.

Older patients with NPM1 mutations also have improved outcomes. As an example, a study from CALGB demonstrated high rates of CR and a three-year OS rate of 35 percent in patients whose blasts were NPM1 mutated and FLT3 wild type [120].

The impact of NPM1 status on selection of post-remission therapy for AML is discussed separately. (See "Post-remission therapy for acute myeloid leukemia in younger adults", section on 'Favorable-risk disease'.)

CEBPA gene — The CEBPA (CCAAT/enhancer binding protein alpha) gene encodes a transcription factor essential for myeloid differentiation [129-131]. CEBPA mutations are one of two known mutation types associated with familial leukemia and can be found in approximately 10 percent of patients with newly diagnosed AML [104,132]. In addition, 13 to 19 percent of patients with cytogenetically normal AML will have CEBPA mutations [52,133-137]. Familial AML with mutated CEBPA has a phenotype that is similar to sporadic AML with biallelic CEBPA mutations. Patients with cytogenetically normal AML with CEBPA mutations have a significantly longer median OS that is independent of other high-risk molecular features [82,132,133,138,139]. (See "Familial disorders of acute leukemia and myelodysplastic syndromes", section on 'Familial AML with mutated CEBPA'.)

As an example, a prospective analysis followed 175 adult patients with newly diagnosed AML with normal cytogenetics who were less than 60 years of age for a median of 4.8 years [132]. Patients with CEBPA mutations had significantly higher rates of five-year EFS (53 versus 30 percent) when compared with those with wild-type CEBPA. In a subset analysis, patients with high-risk molecular features (ie, FLT3-ITD positive and/or wild-type NPM1) who had mutations in CEBPA had significantly better rates of five-year EFS (55 versus 17 percent) and OS (58 versus 27 percent) when compared with those with unmutated CEBPA.

The favorable effect of CEBPA mutations may be limited to patients who carry two copies of the mutant allele and are negative for FLT3-ITD mutations and IDH2 R140 mutations [18,138,140-143]. An international study of 1182 patients with cytogenetically normal AML reported double mutations of CEBPA (either two different mutations or one homozygous mutation) in 91 cases and single mutations in 60 cases [137]. When compared with patients with double mutations of CEBPA, patients with a single CEBPA mutation had higher rates of concurrent mutations in NPM1 (35 versus 3 percent) and FLT3-ITD (30 versus 8 percent). While the presence of any CEBPA mutation was associated with a favorable outcome, only the presence of double mutations of CEBPA was an independent prognostic factor on multivariable analysis. Therefore, the data mentioned above, some of which included patients with only a single mutated allele, may need further study. In addition, there appears to be a separate group of patients in whom CEBPA expression is "silenced" by DNA hypermethylation, the prognostic effect of which is unknown [144].

IDH genes — Somatic mutations in the genes encoding isocitrate dehydrogenase (IDH1 and IDH2) are present in approximately 15 percent of newly diagnosed AML [60-65,104]. IDH1/2 mutations are mutually exclusive with TET2 and WT1 mutations, and are more commonly seen in cases with NPM1 and DNMT3A mutations [104].

Data are conflicting regarding the prognostic impact of IDH gene mutations. In one study, IDH2 mutations appeared to be associated with improved OS, but this benefit was only present in patients with IDH2 R140Q mutations [104]. Concurrent abnormalities in FLT3-ITD appeared to abrogate the beneficial impact of the IDH2 mutation. In another study, patients with IDH2 R172 mutations had a relatively good prognosis, similar to that seen with biallelic CEBPA mutations [18].

Mutant IDH1 can heterodimerize with wild-type IDH1 to create a mutant enzyme that converts alpha-ketoglutarate to 2-hydroxyglutarate (2-HG), which acts as an "oncometabolite" that blocks differentiation [145-148]. IDH1 mutations alone do not appear to be sufficient to induce transformation, but inhibition of mutant IDH1 appears to induce apoptosis and decrease replication in tumor models [149]. In one study, patients with IDH1/2 mutations had significantly increased serum 2-HG levels, and normalization of 2-HG levels after induction therapy was associated with better OS and DFS [150].

Enasidenib, an IDH2 inhibitor, is approved by the US Food and Drug Administration for treatment of patients with relapsed or refractory AML with mutant IDH2 [151]. Inhibitors of IDH1 (eg, AG120, IDH305) and both IDH1 and IDH2 (AG881) are undergoing clinical evaluation in AML and other hematologic malignancies. In particular, AG120 produced a high response rate in patients with relapsed or refractory AML with an IDH1 mutation and hence we also incorporate molecular analysis for IDH1 and IDH2 and NPM1 mutations into the pretreatment evaluation of all patients with newly diagnosed AML and normal karyotypes, regardless of age. (See "Treatment of relapsed or refractory acute myeloid leukemia", section on 'Remission re-induction'.)

KIT gene — Mutations of the KIT gene can be detected in approximately 6 percent of newly diagnosed AML and in 20 to 30 percent of patients with AML and either t(8;21) or inv(16) [104,152,153]. While some studies suggest that KIT gene mutations confer a higher risk of relapse and adversely affect OS in those with inv(16) [152,153], others suggest that this negative prognostic effect is only seen among AML with t(8;21) [104]. Screening for KIT mutations might also allow for use of tyrosine kinase inhibitors such as imatinib or dasatinib, which have in vitro activity against some (but not all) KIT mutations [154]. Clinical trials evaluating the addition of tyrosine kinase inhibitors in selected patients with KIT mutations are in progress.

In one study performed in older patients with high grade MDS and at least 20 percent of myeloblasts expressing KIT, treatment with a combination of imatinib and low dose cytosine arabinoside was unsuccessful [155]. The presence of KIT mutations was not assessed in this study.

WT1 gene — The Wilms tumor 1 gene (WT1) encodes a transcriptional regulator for genes involved in cellular growth and maturation. Disruption of this gene is thought to promote the proliferation of stem cells and disrupt cellular differentiation. Approximately 8 percent of AML cases and 13 percent of patients with cytogenetically normal AML will harbor mutations in WT1 [104,156]. Studies investigating the prognostic value of WT1 gene mutations or single nucleotide polymorphisms in cytogenetically normal AML have had mixed results [68,70,104,156-162]. Some report inferior rates of DFS and OS in patients with WT1 gene mutations while others do not.

ASXL1 and ASXL2 genes — The additional sex combs gene (ASXL1) is a human analog of the Drosophila gene located in chromosome 20q11. Mutations in the ASXL1 gene are present in 6 to 30 percent of cytogenetically normal AML and denote a poor prognosis [18,163-167]. The incidence of ASXL1 mutations in AML increases with age and is higher among those with a history of another myeloid malignancy (eg, myelodysplastic syndrome) [16,167,168].

ASXL1 mutations and NPM1 mutations are mutually exclusive, whereas ASXL1 mutations are strongly associated with alterations in regulators of RNA splicing, such as SRSF2 at 17q25 [18,164,169]. ASXL2 mutations are associated mutations of RUNX1 (also called AML1 or CBFA2) at 21q22 [18,169]. The prognostic impact of mutations in ASXL1 and SRSF2 appears to be additive; while the presence of either portends a poor prognosis, patients with both abnormalities have an even worse prognosis.

The biologic function of ASXL1 is unclear, but may be related to histone post-translational modifications.

DNMT3A gene — The DNMT3A (DNA [cytosine-5]-methyltransferase 3A) gene, located in 2p23.3, plays a role in epigenetic modifications necessary for mammalian development and cell differentiation. Mutations in DNMT3 lead to hypomethylation which, in turn, affects hematopoietic stem cell differentiation. Mutations in the DNMT3A gene are present in 20 to 22 percent of cytogenetically normal AML [79,80,85,86,88,91,170]. However, mutations of DNMT3A and other genes (eg, TET2, ASXL) are found in up to 10 percent of apparently normal adults 65 and older, increasing in incidence with advancing age, referred to as clonal hematopoiesis of indeterminate potential (CHIP) [171-173]. (See "Clonal hematopoiesis of indeterminate potential (CHIP) and related disorders of clonal hematopoiesis".)

Studies have reported mixed effects of DNMT3 mutations on prognosis. While DNMT3A mutations are generally associated with a poor prognosis, its prognostic impact may be affected by coexisting mutations in FLT3, NPM1, and IDH1/2. In one report, the presence of a DNMT3A mutation appeared to have a negative impact on the prognosis of patients without mutation in NPM1 or FLT3, but not of those with NPM1-mutated/FLT3 wild-type AML [91]. In contrast, another report found that DNMT3A mutations were associated with a worse outcome irrespective of the NPM1-mutation status [174]. The interpretation of DNMT3A mutation status is complicated by the high coincidence of DNMT3A mutations and NPM1 mutations (where the two mutations have opposite effects on prognosis) and a potential difference in effect according to the site of DNMT3A mutation.

TP53 gene — Mutations in the TP53 gene are seen in approximately 6 to 8 percent of de novo AML cases, often in cases with complex karyotype or other genetic abnormalities [18,175]. The prognostic impact of TP53 mutation and complex karyotype appears to be additive. Presence of either portends a poor prognosis, while patients with both abnormalities have an even worse prognosis.

Spliceosome mutations — Mutations of regulators of RNA splicing (eg, SRSF2, SF3B1, U2AF1, and ZRSR2) are often associated with mutations of chromatin-modifying genes (ASXL1, STAG2, BCOR, MLL, EZH2, and PHF6) and, together, patients with mutations in these two groups of genes constitute 18 percent of those with newly diagnosed AML [18]. Patients with these mutations are generally older, more frequently have antecedent myeloid disorders (eg, MDS), and have worse clinical outcomes.

Gene expression profiling — There is interest in the use of gene expression profiling (GEP) for the diagnosis, classification, and assessment of prognosis in AML, but it is not yet used routinely in clinical practice [176-186].

Several studies have analyzed leukemia cells from patients with AML and have identified gene "signatures" that may be used to distinguish subsets with different outcomes [177,187-190]. Subgroups with different gene expression profiles have been found in patients with normal cytogenetics [191], as well as those with well-defined cytogenetic changes, such as t(8;21) and inv(16) [192]. Other groups, such as t(15;17), appeared to be more homogeneous in their "signature."

GEP may also be able to identify other groups of AML patients with specific molecular signatures [178,193]. One cluster, associated with the lowest OS and highest cumulative relapse rate after CR, had a high frequency of poor prognostic markers (eg, del(7q), del(5q), t(9;22)). Normal CD34+ cells segregated into this cluster, suggesting that the molecular signature of treatment resistance resembles that of normal hematopoietic stem cells [194]. In another study, AML demonstrating a GEP signature of leukemic stem cells was associated with significantly worse OS, independent of other genetic features [195]. (See "Pathogenesis of acute myeloid leukemia", section on 'Transformation within primitive multipotent cells' and "Pathogenesis of acute myeloid leukemia", section on 'Leukemic stem cells'.)

There was still a wide range of outcomes in the prognostic groupings defined by GEP; accordingly, gene profiling in AML cannot as yet be used as a predictor in individual patients. However, a number of conclusions can be reached from these initial GEP data:

They confirm the importance of cytogenetic subgroups of AML as relatively homogeneous diseases, since leukemias with distinct translocations tend to have very similar gene expression patterns [196].

They begin to subdivide the large group of patients with normal karyotypes into different biological subsets that appear to have different outcomes [197].

They support other data that suggest the role of a leukemic stem cell in the pathogenesis of AML [195].

GEP studies may help to identify critical genes and their protein products whose expression could be modified by available drugs or new agents rationally designed to affect these targets [198].

GEP may also help to define simpler algorithms incorporating a smaller number of genes, which might be detected using more widely available techniques, such as RT-PCR [176,199].

Before these findings can be applied clinically to define subclasses of patients, predict outcome, and perhaps even determine appropriate treatment, they need to be verified in larger groups of patients. In addition, the synthesis and clinical development of drugs to target these heterogeneous molecular changes will be very challenging.

MicroRNA expression profiling — MicroRNA are short sequences of single-stranded RNA that regulate gene expression. The role of microRNA expression profiling is becoming more prominent in our understanding of the pathogenesis of many cancers, including AML [67,200-208].

As an example, a small retrospective study that examined clinical outcomes in higher risk (FLT3 mutated, NPM wild-type) patients with cytogenetically normal AML reported that different patterns of microRNA expression are associated with varying rates of EFS [200]. While this was a small retrospective study that requires further confirmation, microRNA expression profiling analysis may lead to clues permitting treatment with agents selectively affecting specific microRNA targets in the future.

Another study in 187 younger adults (<60 years) with cytogenetically normal AML noted that patients whose tumors had higher expression of a single microRNA (miR-181a) had higher rates of CR and longer OS [209]. MiR-181a expression maintained its prognostic significance in tumors with FLT3-ITD and/or NPM1 wild-type. In contrast, another study of 363 patients with cytogenetically normal AML found that patients whose malignant cells had higher expression of another microRNA (miR-155) were less likely to attain a CR and had OS [210].

Further study is needed to determine how information regarding microRNA expression can be incorporated into clinical practice.

Tumor characteristics — Tumor characteristics can affect patient outcome. Characteristics that have been suggested as prognostic markers include the overexpression of drug efflux pumps, apoptosis inhibitors, antigen expression, or factors that lead to cell cycle progression [11,211-222]. It is unknown how best to integrate this information into clinical practice at this time. Examples follow.

MDR1 phenotype and P-glycoprotein — Overexpression of drug efflux pumps (eg, P-glycoprotein) has been associated with poor outcome in patients with AML, particularly in the elderly [211]. Conflicting observations about the effect of differences in multidrug resistance protein (MRP) expression have been published, perhaps related to the use of different assays. As an example, in one study detection of MRP1 by immunolabeling or RT-PCR failed to show a correlation with treatment outcome, whereas concomitant analysis via a functional assay did show a correlation [212].

The MDR1 phenotype may emerge during the evolutionary process of the leukemic cells. In a report from the SWOG, 71 percent of older patients with AML expressed MDR1, compared with 30 percent in younger subjects [11]. Patients who were MDR1 positive were less likely to have a CR and more likely to have resistant disease. The significance of this finding is unclear, since other studies have indicated that the presence of the MDR-1 phenotype is [213,214] or is not [215] associated with reduced OS in AML [216]. On the other hand, in the SWOG study, older patients who had MDR1-negative AML cells and favorable or intermediate cytogenetics had a high CR rate of 81 percent [11].

In a study involving 153 previously untreated patients with AML, positivity for P-glycoprotein (Pgp) did not adversely affect attainment of CR or OS unless Pgp was expressed along with lung resistance-related protein (LRP) [217]. The mean age of this latter sub-population (LRP+/Pgp+) was 64 years, whereas that of the other groups (LRP+/Pgp-, LRP-/Pgp+, and LRP-/Pgp-) was 48 years, indicating that this adverse prognostic combination was more common in the older patient.

There are a number of inhibitors of PgP mediated drug efflux that have been evaluated in randomized clinical trials. Unfortunately, these studies did not demonstrate improved outcomes in patients receiving the inhibitors in combination with chemotherapy and in some trials, toxicity was increased as well [223].

Overexpression of EVI1 — The ecotropic viral integration site 1 (EVI1) oncogene at chromosome 3q26 is abnormally overexpressed in approximately 8 to 10 percent of AML cases and appears to portend a poor prognosis [218,219,224]. EVI1 appears to interact with DNA methyltransferases that are involved in the epigenetic control of gene expression [225]. (See "Molecular genetics of acute myeloid leukemia", section on 'Mutations affecting DNA methylation'.)

As an example, a study of 1382 adults less than 60 years of age enrolled on multicenter prospective trials evaluated cases for EVI1 expression by polymerase chain reaction (PCR) [219]. EVI1 overexpression was detected in 38, 7, and 0.4 percent of cases with unfavorable, intermediate, and favorable risk cytogenetics, respectively. The prognostic effect of EVI1 overexpression was most apparent in the intermediate-risk group where overexpression was associated with a significantly lower rate of RFS and EFS and a nonsignificant trend toward decreased OS. (See "Cytogenetic abnormalities in acute myeloid leukemia".)

Overexpression of inhibitors of apoptosis — Overexpression of inhibitors of apoptosis, such as BCL2 [220] or survivin [221], may be associated with poorer outcome. High expression of the costimulatory molecule CD40 and the adhesion molecule CD11a have also been associated with a worse outcome [226]. In contrast, overexpression of the apoptosis promoter, BAX, or a high BAX/BCL2 ratio, was associated with better outcome in two studies [227,228]. Many studies are in progress evaluating the use of venetoclax, a BCL2 inhibitor, in AML [229].

CD25 expression — Expression of CD25 (IL-2 receptor alpha) may predict worse outcomes among patients with de novo AML. While initial small retrospective studies suggested that CD25 expression was associated with lower rates of CR and worse OS [230,231], its independent predictive value was questioned since CD25 expression has been shown to correlate with FLT3-ITD gene mutations [231].

The independent prognostic value of CD25 expression in patients with AML was best demonstrated in an analysis of 657 younger adults (≤60 years) with de novo AML treated in a prospective cooperative group trial (ECOG E1900) [232]. Expression of CD25 was identified in 87 patients (13 percent). When compared with CD25 negative tumors, those that express CD25 had inferior rates of CR and OS when stratified by cytogenetic risk group. Among the 75 patients with CD25 expression with mutational analysis results available, 57 (76 percent) demonstrated FLT3-ITD mutations. Among patients with FLT3-ITD mutations, expression of CD25 was associated with shorter median OS (10 versus 25 months) and a lower rate of OS at three years (4 versus 42 percent).

MEASURABLE RESIDUAL DISEASE (MRD) — Measurable residual disease (MRD; also referred to as minimal residual disease) refers to detection of malignant cells, even in the setting of apparent hematologic complete remission. The prognostic value of MRD for AML is presently uncertain. (See "Induction therapy for acute myeloid leukemia in medically-fit adults", section on 'Remission assessment'.)

Methods for detecting MRD in AML include multiparameter flow cytometry, polymerase chain reaction (PCR), and next-generation sequencing. Details of MRD techniques, standardization of assays, types and timing of samples, and other technical aspects of MRD detection are described separately. (See "Induction therapy for acute myeloid leukemia in medically-fit adults".)

Studies that have evaluated the prognostic value of MRD in AML are discussed separately. (See "Induction therapy for acute myeloid leukemia in medically-fit adults", section on 'Remission assessment'.)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient education" and the keyword(s) of interest.)

Beyond the Basics topics (see "Patient education: Acute myeloid leukemia (AML) treatment in adults (Beyond the Basics)")

SUMMARY

The term acute myeloid leukemia (AML) refers to a group of hematopoietic neoplasms involving cells committed to the myeloid line of cellular development. AML is characterized by a clonal proliferation of myeloid precursors with reduced capacity to differentiate into more mature cellular elements.

The response to treatment and overall survival of patients with AML is heterogeneous. A number of prognostic factors related to patient and tumor characteristics have been described for AML (table 1). Of these, patient age at diagnosis, performance status, and karyotype have the most direct effect on treatment at this time and should be a part of the initial evaluation of all patients with newly diagnosed AML. (See 'Clinical risk factors' above and 'Karyotype' above.)

The clinical role of gene mutation analysis, gene expression profiling, and microRNA profiling remains uncertain at this time, although a number of mutations and changes in levels of certain proteins have prognostic impact and increasingly are part of the "routine" characterization of AML (table 4 and table 5 and table 6).

  1. Sekeres MA, Peterson B, Dodge RK, et al. Differences in prognostic factors and outcomes in African Americans and whites with acute myeloid leukemia. Blood 2004; 103:4036.
  2. Olesen LH, Aggerholm A, Andersen BL, et al. Molecular typing of adult acute myeloid leukaemia: significance of translocations, tandem duplications, methylation, and selective gene expression profiling. Br J Haematol 2005; 131:457.
  3. Estey EH. Therapeutic options for acute myelogenous leukemia. Cancer 2001; 92:1059.
  4. Shah A, Andersson TM, Rachet B, et al. Survival and cure of acute myeloid leukaemia in England, 1971-2006: a population-based study. Br J Haematol 2013; 162:509.
  5. Creutzig U, Büchner T, Sauerland MC, et al. Significance of age in acute myeloid leukemia patients younger than 30 years: a common analysis of the pediatric trials AML-BFM 93/98 and the adult trials AMLCG 92/99 and AMLSG HD93/98A. Cancer 2008; 112:562.
  6. Haferlach T, Schoch C, Löffler H, et al. Morphologic dysplasia in de novo acute myeloid leukemia (AML) is related to unfavorable cytogenetics but has no independent prognostic relevance under the conditions of intensive induction therapy: results of a multiparameter analysis from the German AML Cooperative Group studies. J Clin Oncol 2003; 21:256.
  7. Timilshina N, Breunis H, Brandwein JM, et al. Do quality of life or physical function at diagnosis predict short-term outcomes during intensive chemotherapy in AML? Ann Oncol 2014; 25:883.
  8. Appelbaum FR, Gundacker H, Head DR, et al. Age and acute myeloid leukemia. Blood 2006; 107:3481.
  9. Löwenberg B, Ossenkoppele GJ, van Putten W, et al. High-dose daunorubicin in older patients with acute myeloid leukemia. N Engl J Med 2009; 361:1235.
  10. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016; 127:2391.
  11. Leith CP, Kopecky KJ, Godwin J, et al. Acute myeloid leukemia in the elderly: assessment of multidrug resistance (MDR1) and cytogenetics distinguishes biologic subgroups with remarkably distinct responses to standard chemotherapy. A Southwest Oncology Group study. Blood 1997; 89:3323.
  12. Manoharan A, Baker RI, Kyle PW. Low-dose combination chemotherapy for acute myeloid leukemia in elderly patients: a novel approach. Am J Hematol 1997; 55:115.
  13. Wahlin A, Markevärn B, Golovleva I, Nilsson M. Prognostic significance of risk group stratification in elderly patients with acute myeloid leukaemia. Br J Haematol 2001; 115:25.
  14. Letendre L, Noel P, Litzow MR, et al. Treatment of acute myelogenous leukemia in the older patient with attenuated high-dose ara-C. Am J Clin Oncol 1998; 21:142.
  15. Bello C, Yu D, Komrokji RS, et al. Outcomes after induction chemotherapy in patients with acute myeloid leukemia arising from myelodysplastic syndrome. Cancer 2011; 117:1463.
  16. Lindsley RC, Mar BG, Mazzola E, et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood 2015; 125:1367.
  17. Döhner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017; 129:424.
  18. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N Engl J Med 2016; 374:2209.
  19. Pastore F, Dufour A, Benthaus T, et al. Combined molecular and clinical prognostic index for relapse and survival in cytogenetically normal acute myeloid leukemia. J Clin Oncol 2014; 32:1586.
  20. Ferrant A, Doyen C, Delannoy A, et al. Karyotype in acute myeloblastic leukemia: prognostic significance in a prospective study assessing bone marrow transplantation in first remission. Bone Marrow Transplant 1995; 15:685.
  21. Keating MJ, Smith TL, Kantarjian H, et al. Cytogenetic pattern in acute myelogenous leukemia: a major reproducible determinant of outcome. Leukemia 1988; 2:403.
  22. Bloomfield CD, Lawrence D, Byrd JC, et al. Frequency of prolonged remission duration after high-dose cytarabine intensification in acute myeloid leukemia varies by cytogenetic subtype. Cancer Res 1998; 58:4173.
  23. Slovak ML, Kopecky KJ, Cassileth PA, et al. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study. Blood 2000; 96:4075.
  24. Byrd JC, Mrózek K, Dodge RK, et al. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461). Blood 2002; 100:4325.
  25. Delaunay J, Vey N, Leblanc T, et al. Prognosis of inv(16)/t(16;16) acute myeloid leukemia (AML): a survey of 110 cases from the French AML Intergroup. Blood 2003; 102:462.
  26. Schlenk RF, Benner A, Krauter J, et al. Individual patient data-based meta-analysis of patients aged 16 to 60 years with core binding factor acute myeloid leukemia: a survey of the German Acute Myeloid Leukemia Intergroup. J Clin Oncol 2004; 22:3741.
  27. Grimwade D, Hills RK, Moorman AV, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood 2010; 116:354.
  28. Grimwade D, Walker H, Oliver F, et al. The importance of diagnostic cytogenetics on outcome in AML: analysis of 1,612 patients entered into the MRC AML 10 trial. The Medical Research Council Adult and Children's Leukaemia Working Parties. Blood 1998; 92:2322.
  29. Wheatley K, Burnett AK, Goldstone AH, et al. A simple, robust, validated and highly predictive index for the determination of risk-directed therapy in acute myeloid leukaemia derived from the MRC AML 10 trial. United Kingdom Medical Research Council's Adult and Childhood Leukaemia Working Parties. Br J Haematol 1999; 107:69.
  30. Appelbaum FR, Kopecky KJ, Tallman MS, et al. The clinical spectrum of adult acute myeloid leukaemia associated with core binding factor translocations. Br J Haematol 2006; 135:165.
  31. Breems DA, Van Putten WL, De Greef GE, et al. Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype. J Clin Oncol 2008; 26:4791.
  32. Kayser S, Zucknick M, Döhner K, et al. Monosomal karyotype in adult acute myeloid leukemia: prognostic impact and outcome after different treatment strategies. Blood 2012; 119:551.
  33. Middeke JM, Beelen D, Stadler M, et al. Outcome of high-risk acute myeloid leukemia after allogeneic hematopoietic cell transplantation: negative impact of abnl(17p) and -5/5q-. Blood 2012; 120:2521.
  34. Medeiros BC, Othus M, Fang M, et al. Prognostic impact of monosomal karyotype in young adult and elderly acute myeloid leukemia: the Southwest Oncology Group (SWOG) experience. Blood 2010; 116:2224.
  35. Fang M, Storer B, Estey E, et al. Outcome of patients with acute myeloid leukemia with monosomal karyotype who undergo hematopoietic cell transplantation. Blood 2011; 118:1490.
  36. Cornelissen JJ, Breems D, van Putten WL, et al. Comparative analysis of the value of allogeneic hematopoietic stem-cell transplantation in acute myeloid leukemia with monosomal karyotype versus other cytogenetic risk categories. J Clin Oncol 2012; 30:2140.
  37. Moorman AV, Hagemeijer A, Charrin C, et al. The translocations, t(11;19)(q23;p13.1) and t(11;19)(q23;p13.3): a cytogenetic and clinical profile of 53 patients. European 11q23 Workshop participants. Leukemia 1998; 12:805.
  38. Swansbury GJ, Slater R, Bain BJ, et al. Hematological malignancies with t(9;11)(p21-22;q23)--a laboratory and clinical study of 125 cases. European 11q23 Workshop participants. Leukemia 1998; 12:792.
  39. Krauter J, Wagner K, Schäfer I, et al. Prognostic factors in adult patients up to 60 years old with acute myeloid leukemia and translocations of chromosome band 11q23: individual patient data-based meta-analysis of the German Acute Myeloid Leukemia Intergroup. J Clin Oncol 2009; 27:3000.
  40. Forrest DL, Nevill TJ, Horsman DE, et al. Bone marrow transplantation for adults with acute leukaemia and 11q23 chromosomal abnormalities. Br J Haematol 1998; 103:630.
  41. Samuels BL, Larson RA, Le Beau MM, et al. Specific chromosomal abnormalities in acute nonlymphocytic leukemia correlate with drug susceptibility in vivo. Leukemia 1988; 2:79.
  42. Baer MR, Stewart CC, Lawrence D, et al. Acute myeloid leukemia with 11q23 translocations: myelomonocytic immunophenotype by multiparameter flow cytometry. Leukemia 1998; 12:317.
  43. Byrd JC, Lawrence D, Arthur DC, et al. Patients with isolated trisomy 8 in acute myeloid leukemia are not cured with cytarabine-based chemotherapy: results from Cancer and Leukemia Group B 8461. Clin Cancer Res 1998; 4:1235.
  44. Wolman SR, Gundacker H, Appelbaum FR, et al. Impact of trisomy 8 (+8) on clinical presentation, treatment response, and survival in acute myeloid leukemia: a Southwest Oncology Group study. Blood 2002; 100:29.
  45. Grimwade D, Walker H, Harrison G, et al. The predictive value of hierarchical cytogenetic classification in older adults with acute myeloid leukemia (AML): analysis of 1065 patients entered into the United Kingdom Medical Research Council AML11 trial. Blood 2001; 98:1312.
  46. Schoch C, Haferlach T, Haase D, et al. Patients with de novo acute myeloid leukaemia and complex karyotype aberrations show a poor prognosis despite intensive treatment: a study of 90 patients. Br J Haematol 2001; 112:118.
  47. Fröhling S, Schlenk RF, Kayser S, et al. Cytogenetics and age are major determinants of outcome in intensively treated acute myeloid leukemia patients older than 60 years: results from AMLSG trial AML HD98-B. Blood 2006; 108:3280.
  48. Knipp S, Hildebrand B, Kündgen A, et al. Intensive chemotherapy is not recommended for patients aged >60 years who have myelodysplastic syndromes or acute myeloid leukemia with high-risk karyotypes. Cancer 2007; 110:345.
  49. Cancer and Leukemia Group B 8461, Farag SS, Archer KJ, et al. Pretreatment cytogenetics add to other prognostic factors predicting complete remission and long-term outcome in patients 60 years of age or older with acute myeloid leukemia: results from Cancer and Leukemia Group B 8461. Blood 2006; 108:63.
  50. Santamaría CM, Chillón MC, García-Sanz R, et al. Molecular stratification model for prognosis in cytogenetically normal acute myeloid leukemia. Blood 2009; 114:148.
  51. Marcucci G, Mrózek K, Bloomfield CD. Molecular heterogeneity and prognostic biomarkers in adults with acute myeloid leukemia and normal cytogenetics. Curr Opin Hematol 2005; 12:68.
  52. Bienz M, Ludwig M, Leibundgut EO, et al. Risk assessment in patients with acute myeloid leukemia and a normal karyotype. Clin Cancer Res 2005; 11:1416.
  53. Marcucci G, Baldus CD, Ruppert AS, et al. Overexpression of the ETS-related gene, ERG, predicts a worse outcome in acute myeloid leukemia with normal karyotype: a Cancer and Leukemia Group B study. J Clin Oncol 2005; 23:9234.
  54. Monzo M, Brunet S, Urbano-Ispizua A, et al. Genomic polymorphisms provide prognostic information in intermediate-risk acute myeloblastic leukemia. Blood 2006; 107:4871.
  55. Pasqualucci L, Liso A, Martelli MP, et al. Mutated nucleophosmin detects clonal multilineage involvement in acute myeloid leukemia: Impact on WHO classification. Blood 2006; 108:4146.
  56. Marcucci G, Maharry K, Whitman SP, et al. High expression levels of the ETS-related gene, ERG, predict adverse outcome and improve molecular risk-based classification of cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B Study. J Clin Oncol 2007; 25:3337.
  57. Langer C, Radmacher MD, Ruppert AS, et al. High BAALC expression associates with other molecular prognostic markers, poor outcome, and a distinct gene-expression signature in cytogenetically normal patients younger than 60 years with acute myeloid leukemia: a Cancer and Leukemia Group B (CALGB) study. Blood 2008; 111:5371.
  58. Andreeff M, Ruvolo V, Gadgil S, et al. HOX expression patterns identify a common signature for favorable AML. Leukemia 2008; 22:2041.
  59. Metzeler KH, Dufour A, Benthaus T, et al. ERG expression is an independent prognostic factor and allows refined risk stratification in cytogenetically normal acute myeloid leukemia: a comprehensive analysis of ERG, MN1, and BAALC transcript levels using oligonucleotide microarrays. J Clin Oncol 2009; 27:5031.
  60. Marcucci G, Maharry K, Wu YZ, et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol 2010; 28:2348.
  61. Wagner K, Damm F, Göhring G, et al. Impact of IDH1 R132 mutations and an IDH1 single nucleotide polymorphism in cytogenetically normal acute myeloid leukemia: SNP rs11554137 is an adverse prognostic factor. J Clin Oncol 2010; 28:2356.
  62. Paschka P, Schlenk RF, Gaidzik VI, et al. IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication. J Clin Oncol 2010; 28:3636.
  63. Abbas S, Lugthart S, Kavelaars FG, et al. Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value. Blood 2010; 116:2122.
  64. Green CL, Evans CM, Hills RK, et al. The prognostic significance of IDH1 mutations in younger adult patients with acute myeloid leukemia is dependent on FLT3/ITD status. Blood 2010; 116:2779.
  65. Schnittger S, Haferlach C, Ulke M, et al. IDH1 mutations are detected in 6.6% of 1414 AML patients and are associated with intermediate risk karyotype and unfavorable prognosis in adults younger than 60 years and unmutated NPM1 status. Blood 2010; 116:5486.
  66. Schwind S, Marcucci G, Maharry K, et al. BAALC and ERG expression levels are associated with outcome and distinct gene and microRNA expression profiles in older patients with de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. Blood 2010; 116:5660.
  67. Eisfeld AK, Marcucci G, Maharry K, et al. miR-3151 interplays with its host gene BAALC and independently affects outcome of patients with cytogenetically normal acute myeloid leukemia. Blood 2012; 120:249.
  68. Paschka P, Marcucci G, Ruppert AS, et al. Wilms' tumor 1 gene mutations independently predict poor outcome in adults with cytogenetically normal acute myeloid leukemia: a cancer and leukemia group B study. J Clin Oncol 2008; 26:4595.
  69. Neubauer A, Maharry K, Mrózek K, et al. Patients with acute myeloid leukemia and RAS mutations benefit most from postremission high-dose cytarabine: a Cancer and Leukemia Group B study. J Clin Oncol 2008; 26:4603.
  70. Virappane P, Gale R, Hills R, et al. Mutation of the Wilms' tumor 1 gene is a poor prognostic factor associated with chemotherapy resistance in normal karyotype acute myeloid leukemia: the United Kingdom Medical Research Council Adult Leukaemia Working Party. J Clin Oncol 2008; 26:5429.
  71. Heuser M, Beutel G, Krauter J, et al. High meningioma 1 (MN1) expression as a predictor for poor outcome in acute myeloid leukemia with normal cytogenetics. Blood 2006; 108:3898.
  72. Summers K, Stevens J, Kakkas I, et al. Wilms' tumour 1 mutations are associated with FLT3-ITD and failure of standard induction chemotherapy in patients with normal karyotype AML. Leukemia 2007; 21:550.
  73. Hollink IH, van den Heuvel-Eibrink MM, Zimmermann M, et al. Clinical relevance of Wilms tumor 1 gene mutations in childhood acute myeloid leukemia. Blood 2009; 113:5951.
  74. Langer C, Marcucci G, Holland KB, et al. Prognostic importance of MN1 transcript levels, and biologic insights from MN1-associated gene and microRNA expression signatures in cytogenetically normal acute myeloid leukemia: a cancer and leukemia group B study. J Clin Oncol 2009; 27:3198.
  75. Tang JL, Hou HA, Chen CY, et al. AML1/RUNX1 mutations in 470 adult patients with de novo acute myeloid leukemia: prognostic implication and interaction with other gene alterations. Blood 2009; 114:5352.
  76. Schnittger S, Dicker F, Kern W, et al. RUNX1 mutations are frequent in de novo AML with noncomplex karyotype and confer an unfavorable prognosis. Blood 2011; 117:2348.
  77. Gaidzik VI, Bullinger L, Schlenk RF, et al. RUNX1 mutations in acute myeloid leukemia: results from a comprehensive genetic and clinical analysis from the AML study group. J Clin Oncol 2011; 29:1364.
  78. Metzeler KH, Maharry K, Radmacher MD, et al. TET2 mutations improve the new European LeukemiaNet risk classification of acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol 2011; 29:1373.
  79. Yan XJ, Xu J, Gu ZH, et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nat Genet 2011; 43:309.
  80. Thol F, Damm F, Lüdeking A, et al. Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia. J Clin Oncol 2011; 29:2889.
  81. Green CL, Evans CM, Zhao L, et al. The prognostic significance of IDH2 mutations in AML depends on the location of the mutation. Blood 2011; 118:409.
  82. Rockova V, Abbas S, Wouters BJ, et al. Risk stratification of intermediate-risk acute myeloid leukemia: integrative analysis of a multitude of gene mutation and gene expression markers. Blood 2011; 118:1069.
  83. Chou WC, Chou SC, Liu CY, et al. TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics. Blood 2011; 118:3803.
  84. Shen Y, Zhu YM, Fan X, et al. Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia. Blood 2011; 118:5593.
  85. Hou HA, Kuo YY, Liu CY, et al. DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications. Blood 2012; 119:559.
  86. Marcucci G, Metzeler KH, Schwind S, et al. Age-related prognostic impact of different types of DNMT3A mutations in adults with primary cytogenetically normal acute myeloid leukemia. J Clin Oncol 2012; 30:742.
  87. Gaidzik VI, Paschka P, Späth D, et al. TET2 mutations in acute myeloid leukemia (AML): results from a comprehensive genetic and clinical analysis of the AML study group. J Clin Oncol 2012; 30:1350.
  88. Ribeiro AF, Pratcorona M, Erpelinck-Verschueren C, et al. Mutant DNMT3A: a marker of poor prognosis in acute myeloid leukemia. Blood 2012; 119:5824.
  89. Mendler JH, Maharry K, Radmacher MD, et al. RUNX1 mutations are associated with poor outcome in younger and older patients with cytogenetically normal acute myeloid leukemia and with distinct gene and MicroRNA expression signatures. J Clin Oncol 2012; 30:3109.
  90. Grossmann V, Schnittger S, Kohlmann A, et al. A novel hierarchical prognostic model of AML solely based on molecular mutations. Blood 2012; 120:2963.
  91. Gaidzik VI, Schlenk RF, Paschka P, et al. Clinical impact of DNMT3A mutations in younger adult patients with acute myeloid leukemia: results of the AML Study Group (AMLSG). Blood 2013; 121:4769.
  92. Metzeler KH, Herold T, Rothenberg-Thurley M, et al. Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia. Blood 2016; 128:686.
  93. Ng SW, Mitchell A, Kennedy JA, et al. A 17-gene stemness score for rapid determination of risk in acute leukaemia. Nature 2016; 540:433.
  94. Schnittger S, Schoch C, Dugas M, et al. Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease. Blood 2002; 100:59.
  95. Kottaridis PD, Gale RE, Linch DC. Flt3 mutations and leukaemia. Br J Haematol 2003; 122:523.
  96. Whitman SP, Archer KJ, Feng L, et al. Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: a cancer and leukemia group B study. Cancer Res 2001; 61:7233.
  97. Fröhling S, Schlenk RF, Breitruck J, et al. Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: a study of the AML Study Group Ulm. Blood 2002; 100:4372.
  98. Zwaan CM, Meshinchi S, Radich JP, et al. FLT3 internal tandem duplication in 234 children with acute myeloid leukemia: prognostic significance and relation to cellular drug resistance. Blood 2003; 102:2387.
  99. Pollard JA, Alonzo TA, Gerbing RB, et al. FLT3 internal tandem duplication in CD34+/CD33- precursors predicts poor outcome in acute myeloid leukemia. Blood 2006; 108:2764.
  100. Artz A, Dong X. Defining anemia by race using epidemiologic data. Blood 2008; 111:2941; author reply 2941.
  101. Schlenk RF, Döhner K, Krauter J, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med 2008; 358:1909.
  102. Kayser S, Schlenk RF, Londono MC, et al. Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome. Blood 2009; 114:2386.
  103. Whitman SP, Maharry K, Radmacher MD, et al. FLT3 internal tandem duplication associates with adverse outcome and gene- and microRNA-expression signatures in patients 60 years of age or older with primary cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. Blood 2010; 116:3622.
  104. Patel JP, Gönen M, Figueroa ME, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med 2012; 366:1079.
  105. Schmid C, Labopin M, Socié G, et al. Outcome of patients with distinct molecular genotypes and cytogenetically normal AML after allogeneic transplantation. Blood 2015; 126:2062.
  106. Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood 2001; 98:1752.
  107. Meshinchi S, Alonzo TA, Stirewalt DL, et al. Clinical implications of FLT3 mutations in pediatric AML. Blood 2006; 108:3654.
  108. Schneider F, Hoster E, Unterhalt M, et al. The FLT3ITD mRNA level has a high prognostic impact in NPM1 mutated, but not in NPM1 unmutated, AML with a normal karyotype. Blood 2012; 119:4383.
  109. Yamamoto Y, Kiyoi H, Nakano Y, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood 2001; 97:2434.
  110. Bacher U, Haferlach C, Kern W, et al. Prognostic relevance of FLT3-TKD mutations in AML: the combination matters--an analysis of 3082 patients. Blood 2008; 111:2527.
  111. Borthakur G, Kantarjian H, Patel KP, et al. Impact of numerical variation in FMS-like tyrosine kinase receptor 3 internal tandem duplications on clinical outcome in normal karyotype acute myelogenous leukemia. Cancer 2012; 118:5819.
  112. Thiede C, Steudel C, Mohr B, et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood 2002; 99:4326.
  113. Gale RE, Green C, Allen C, et al. The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood 2008; 111:2776.
  114. Döhner K, Thiede C, Jahn N, et al. Impact of NPM1/FLT3-ITD genotypes defined by the 2017 European LeukemiaNet in patients with acute myeloid leukemia. Blood 2020; 135:371.
  115. Gale RE, Hills R, Kottaridis PD, et al. No evidence that FLT3 status should be considered as an indicator for transplantation in acute myeloid leukemia (AML): an analysis of 1135 patients, excluding acute promyelocytic leukemia, from the UK MRC AML10 and 12 trials. Blood 2005; 106:3658.
  116. Brunet S, Labopin M, Esteve J, et al. Impact of FLT3 internal tandem duplication on the outcome of related and unrelated hematopoietic transplantation for adult acute myeloid leukemia in first remission: a retrospective analysis. J Clin Oncol 2012; 30:735.
  117. Falini B, Nicoletti I, Martelli MF, Mecucci C. Acute myeloid leukemia carrying cytoplasmic/mutated nucleophosmin (NPMc+ AML): biologic and clinical features. Blood 2007; 109:874.
  118. Hollink IH, Zwaan CM, Zimmermann M, et al. Favorable prognostic impact of NPM1 gene mutations in childhood acute myeloid leukemia, with emphasis on cytogenetically normal AML. Leukemia 2009; 23:262.
  119. Schneider F, Hoster E, Unterhalt M, et al. NPM1 but not FLT3-ITD mutations predict early blast cell clearance and CR rate in patients with normal karyotype AML (NK-AML) or high-risk myelodysplastic syndrome (MDS). Blood 2009; 113:5250.
  120. Becker H, Marcucci G, Maharry K, et al. Favorable prognostic impact of NPM1 mutations in older patients with cytogenetically normal de novo acute myeloid leukemia and associated gene- and microRNA-expression signatures: a Cancer and Leukemia Group B study. J Clin Oncol 2010; 28:596.
  121. Falini B, Martelli MP, Bolli N, et al. Acute myeloid leukemia with mutated nucleophosmin (NPM1): is it a distinct entity? Blood 2011; 117:1109.
  122. Haferlach C, Mecucci C, Schnittger S, et al. AML with mutated NPM1 carrying a normal or aberrant karyotype show overlapping biologic, pathologic, immunophenotypic, and prognostic features. Blood 2009; 114:3024.
  123. Patel SS, Kuo FC, Gibson CJ, et al. High NPM1-mutant allele burden at diagnosis predicts unfavorable outcomes in de novo AML. Blood 2018; 131:2816.
  124. Angenendt L, Röllig C, Montesinos P, et al. Chromosomal Abnormalities and Prognosis in NPM1-Mutated Acute Myeloid Leukemia: A Pooled Analysis of Individual Patient Data From Nine International Cohorts. J Clin Oncol 2019; 37:2632.
  125. Döhner K, Schlenk RF, Habdank M, et al. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations. Blood 2005; 106:3740.
  126. Verhaak RG, Goudswaard CS, van Putten W, et al. Mutations in nucleophosmin (NPM1) in acute myeloid leukemia (AML): association with other gene abnormalities and previously established gene expression signatures and their favorable prognostic significance. Blood 2005; 106:3747.
  127. Thiede C, Koch S, Creutzig E, et al. Prevalence and prognostic impact of NPM1 mutations in 1485 adult patients with acute myeloid leukemia (AML). Blood 2006; 107:4011.
  128. Falini B, Martelli MP, Bolli N, et al. Immunohistochemistry predicts nucleophosmin (NPM) mutations in acute myeloid leukemia. Blood 2006; 108:1999.
  129. Nerlov C. C/EBPalpha mutations in acute myeloid leukaemias. Nat Rev Cancer 2004; 4:394.
  130. Eyholzer M, Schmid S, Wilkens L, et al. The tumour-suppressive miR-29a/b1 cluster is regulated by CEBPA and blocked in human AML. Br J Cancer 2010; 103:275.
  131. Avellino R, Havermans M, Erpelinck C, et al. An autonomous CEBPA enhancer specific for myeloid-lineage priming and neutrophilic differentiation. Blood 2016; 127:2991.
  132. Marcucci G, Maharry K, Radmacher MD, et al. Prognostic significance of, and gene and microRNA expression signatures associated with, CEBPA mutations in cytogenetically normal acute myeloid leukemia with high-risk molecular features: a Cancer and Leukemia Group B Study. J Clin Oncol 2008; 26:5078.
  133. Fröhling S, Schlenk RF, Stolze I, et al. CEBPA mutations in younger adults with acute myeloid leukemia and normal cytogenetics: prognostic relevance and analysis of cooperating mutations. J Clin Oncol 2004; 22:624.
  134. Pabst T, Eyholzer M, Haefliger S, et al. Somatic CEBPA mutations are a frequent second event in families with germline CEBPA mutations and familial acute myeloid leukemia. J Clin Oncol 2008; 26:5088.
  135. Boissel N, Renneville A, Biggio V, et al. Prevalence, clinical profile, and prognosis of NPM mutations in AML with normal karyotype. Blood 2005; 106:3618.
  136. Ho PA, Alonzo TA, Gerbing RB, et al. Prevalence and prognostic implications of CEBPA mutations in pediatric acute myeloid leukemia (AML): a report from the Children's Oncology Group. Blood 2009; 113:6558.
  137. Taskesen E, Bullinger L, Corbacioglu A, et al. Prognostic impact, concurrent genetic mutations, and gene expression features of AML with CEBPA mutations in a cohort of 1182 cytogenetically normal AML patients: further evidence for CEBPA double mutant AML as a distinctive disease entity. Blood 2011; 117:2469.
  138. Renneville A, Boissel N, Gachard N, et al. The favorable impact of CEBPA mutations in patients with acute myeloid leukemia is only observed in the absence of associated cytogenetic abnormalities and FLT3 internal duplication. Blood 2009; 113:5090.
  139. Bacher U, Schnittger S, Macijewski K, et al. Multilineage dysplasia does not influence prognosis in CEBPA-mutated AML, supporting the WHO proposal to classify these patients as a unique entity. Blood 2012; 119:4719.
  140. Wouters BJ, Löwenberg B, Erpelinck-Verschueren CA, et al. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood 2009; 113:3088.
  141. Pabst T, Eyholzer M, Fos J, Mueller BU. Heterogeneity within AML with CEBPA mutations; only CEBPA double mutations, but not single CEBPA mutations are associated with favourable prognosis. Br J Cancer 2009; 100:1343.
  142. Dufour A, Schneider F, Metzeler KH, et al. Acute myeloid leukemia with biallelic CEBPA gene mutations and normal karyotype represents a distinct genetic entity associated with a favorable clinical outcome. J Clin Oncol 2010; 28:570.
  143. Green CL, Koo KK, Hills RK, et al. Prognostic significance of CEBPA mutations in a large cohort of younger adult patients with acute myeloid leukemia: impact of double CEBPA mutations and the interaction with FLT3 and NPM1 mutations. J Clin Oncol 2010; 28:2739.
  144. Figueroa ME, Wouters BJ, Skrabanek L, et al. Genome-wide epigenetic analysis delineates a biologically distinct immature acute leukemia with myeloid/T-lymphoid features. Blood 2009; 113:2795.
  145. Dang L, White DW, Gross S, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 2009; 462:739.
  146. Xu W, Yang H, Liu Y, et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 2011; 19:17.
  147. Pietrak B, Zhao H, Qi H, et al. A tale of two subunits: how the neomorphic R132H IDH1 mutation enhances production of αHG. Biochemistry 2011; 50:4804.
  148. Losman JA, Looper RE, Koivunen P, et al. (R)-2-hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible. Science 2013; 339:1621.
  149. Chaturvedi A, Araujo Cruz MM, Jyotsana N, et al. Mutant IDH1 promotes leukemogenesis in vivo and can be specifically targeted in human AML. Blood 2013; 122:2877.
  150. Janin M, Mylonas E, Saada V, et al. Serum 2-hydroxyglutarate production in IDH1- and IDH2-mutated de novo acute myeloid leukemia: a study by the Acute Leukemia French Association group. J Clin Oncol 2014; 32:297.
  151. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017; 130:722.
  152. Paschka P, Marcucci G, Ruppert AS, et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): a Cancer and Leukemia Group B Study. J Clin Oncol 2006; 24:3904.
  153. Lück SC, Russ AC, Du J, et al. KIT mutations confer a distinct gene expression signature in core binding factor leukaemia. Br J Haematol 2010; 148:925.
  154. Kolitz JE. Current therapeutic strategies for acute myeloid leukaemia. Br J Haematol 2006; 134:555.
  155. Heidel F, Cortes J, Rücker FG, et al. Results of a multicenter phase II trial for older patients with c-Kit-positive acute myeloid leukemia (AML) and high-risk myelodysplastic syndrome (HR-MDS) using low-dose Ara-C and Imatinib. Cancer 2007; 109:907.
  156. Gaidzik VI, Schlenk RF, Moschny S, et al. Prognostic impact of WT1 mutations in cytogenetically normal acute myeloid leukemia: a study of the German-Austrian AML Study Group. Blood 2009; 113:4505.
  157. Renneville A, Boissel N, Zurawski V, et al. Wilms tumor 1 gene mutations are associated with a higher risk of recurrence in young adults with acute myeloid leukemia: a study from the Acute Leukemia French Association. Cancer 2009; 115:3719.
  158. Cilloni D, Renneville A, Hermitte F, et al. Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia: a European LeukemiaNet study. J Clin Oncol 2009; 27:5195.
  159. Damm F, Heuser M, Morgan M, et al. Single nucleotide polymorphism in the mutational hotspot of WT1 predicts a favorable outcome in patients with cytogenetically normal acute myeloid leukemia. J Clin Oncol 2010; 28:578.
  160. Hou HA, Huang TC, Lin LI, et al. WT1 mutation in 470 adult patients with acute myeloid leukemia: stability during disease evolution and implication of its incorporation into a survival scoring system. Blood 2010; 115:5222.
  161. Ho PA, Zeng R, Alonzo TA, et al. Prevalence and prognostic implications of WT1 mutations in pediatric acute myeloid leukemia (AML): a report from the Children's Oncology Group. Blood 2010; 116:702.
  162. Becker H, Marcucci G, Maharry K, et al. Mutations of the Wilms tumor 1 gene (WT1) in older patients with primary cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. Blood 2010; 116:788.
  163. Abdel-Wahab O, Manshouri T, Patel J, et al. Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. Cancer Res 2010; 70:447.
  164. Carbuccia N, Trouplin V, Gelsi-Boyer V, et al. Mutual exclusion of ASXL1 and NPM1 mutations in a series of acute myeloid leukemias. Leukemia 2010; 24:469.
  165. Boultwood J, Perry J, Pellagatti A, et al. Frequent mutation of the polycomb-associated gene ASXL1 in the myelodysplastic syndromes and in acute myeloid leukemia. Leukemia 2010; 24:1062.
  166. Chou WC, Huang HH, Hou HA, et al. Distinct clinical and biological features of de novo acute myeloid leukemia with additional sex comb-like 1 (ASXL1) mutations. Blood 2010; 116:4086.
  167. Schnittger S, Eder C, Jeromin S, et al. ASXL1 exon 12 mutations are frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome. Leukemia 2013; 27:82.
  168. Metzeler KH, Becker H, Maharry K, et al. ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN Favorable genetic category. Blood 2011; 118:6920.
  169. Micol JB, Duployez N, Boissel N, et al. Frequent ASXL2 mutations in acute myeloid leukemia patients with t(8;21)/RUNX1-RUNX1T1 chromosomal translocations. Blood 2014; 124:1445.
  170. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med 2010; 363:2424.
  171. Xie M, Lu C, Wang J, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med 2014; 20:1472.
  172. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 2014; 371:2488.
  173. Genovese G, Kähler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 2014; 371:2477.
  174. Gale RE, Lamb K, Allen C, et al. Simpson's Paradox and the Impact of Different DNMT3A Mutations on Outcome in Younger Adults With Acute Myeloid Leukemia. J Clin Oncol 2015; 33:2072.
  175. Cancer Genome Atlas Research Network, Ley TJ, Miller C, et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 2013; 368:2059.
  176. Lacayo NJ, Meshinchi S, Kinnunen P, et al. Gene expression profiles at diagnosis in de novo childhood AML patients identify FLT3 mutations with good clinical outcomes. Blood 2004; 104:2646.
  177. Bullinger L, Döhner K, Bair E, et al. Use of gene-expression profiling to identify prognostic subclasses in adult acute myeloid leukemia. N Engl J Med 2004; 350:1605.
  178. Valk PJ, Verhaak RG, Beijen MA, et al. Prognostically useful gene-expression profiles in acute myeloid leukemia. N Engl J Med 2004; 350:1617.
  179. Wilson CS, Davidson GS, Martin SB, et al. Gene expression profiling of adult acute myeloid leukemia identifies novel biologic clusters for risk classification and outcome prediction. Blood 2006; 108:685.
  180. Rücker FG, Bullinger L, Schwaenen C, et al. Disclosure of candidate genes in acute myeloid leukemia with complex karyotypes using microarray-based molecular characterization. J Clin Oncol 2006; 24:3887.
  181. Hess CJ, Berkhof J, Denkers F, et al. Activated intrinsic apoptosis pathway is a key related prognostic parameter in acute myeloid leukemia. J Clin Oncol 2007; 25:1209.
  182. Bullinger L, Döhner K, Kranz R, et al. An FLT3 gene-expression signature predicts clinical outcome in normal karyotype AML. Blood 2008; 111:4490.
  183. Bacher U, Kohlmann A, Haferlach T. Current status of gene expression profiling in the diagnosis and management of acute leukaemia. Br J Haematol 2009; 145:555.
  184. Powell JA, Thomas D, Barry EF, et al. Expression profiling of a hemopoietic cell survival transcriptome implicates osteopontin as a functional prognostic factor in AML. Blood 2009; 114:4859.
  185. Haferlach T, Kohlmann A, Wieczorek L, et al. Clinical utility of microarray-based gene expression profiling in the diagnosis and subclassification of leukemia: report from the International Microarray Innovations in Leukemia Study Group. J Clin Oncol 2010; 28:2529.
  186. Gentles AJ, Plevritis SK, Majeti R, Alizadeh AA. Association of a leukemic stem cell gene expression signature with clinical outcomes in acute myeloid leukemia. JAMA 2010; 304:2706.
  187. Metzeler KH, Hummel M, Bloomfield CD, et al. An 86-probe-set gene-expression signature predicts survival in cytogenetically normal acute myeloid leukemia. Blood 2008; 112:4193.
  188. Silva FP, Swagemakers SM, Erpelinck-Verschueren C, et al. Gene expression profiling of minimally differentiated acute myeloid leukemia: M0 is a distinct entity subdivided by RUNX1 mutation status. Blood 2009; 114:3001.
  189. de Jonge HJ, Valk PJ, Veeger NJ, et al. High VEGFC expression is associated with unique gene expression profiles and predicts adverse prognosis in pediatric and adult acute myeloid leukemia. Blood 2010; 116:1747.
  190. Li Z, Herold T, He C, et al. Identification of a 24-gene prognostic signature that improves the European LeukemiaNet risk classification of acute myeloid leukemia: an international collaborative study. J Clin Oncol 2013; 31:1172.
  191. Radmacher MD, Marcucci G, Ruppert AS, et al. Independent confirmation of a prognostic gene-expression signature in adult acute myeloid leukemia with a normal karyotype: a Cancer and Leukemia Group B study. Blood 2006; 108:1677.
  192. Bullinger L, Rücker FG, Kurz S, et al. Gene-expression profiling identifies distinct subclasses of core binding factor acute myeloid leukemia. Blood 2007; 110:1291.
  193. de Jonge HJ, de Bont ES, Valk PJ, et al. AML at older age: age-related gene expression profiles reveal a paradoxical down-regulation of p16INK4A mRNA with prognostic significance. Blood 2009; 114:2869.
  194. van Rhenen A, Feller N, Kelder A, et al. High stem cell frequency in acute myeloid leukemia at diagnosis predicts high minimal residual disease and poor survival. Clin Cancer Res 2005; 11:6520.
  195. Greaves M. Cancer stem cells renew their impact. Nat Med 2011; 17:1046.
  196. Haferlach T, Kohlmann A, Schnittger S, et al. Global approach to the diagnosis of leukemia using gene expression profiling. Blood 2005; 106:1189.
  197. Mrózek K, Marcucci G, Paschka P, et al. Clinical relevance of mutations and gene-expression changes in adult acute myeloid leukemia with normal cytogenetics: are we ready for a prognostically prioritized molecular classification? Blood 2007; 109:431.
  198. Tallman MS, Gilliland DG, Rowe JM. Drug therapy for acute myeloid leukemia. Blood 2005; 106:1154.
  199. Sakhinia E, Faranghpour M, Liu Yin JA, et al. Routine expression profiling of microarray gene signatures in acute leukaemia by real-time PCR of human bone marrow. Br J Haematol 2005; 130:233.
  200. Marcucci G, Radmacher MD, Maharry K, et al. MicroRNA expression in cytogenetically normal acute myeloid leukemia. N Engl J Med 2008; 358:1919.
  201. Garzon R, Volinia S, Liu CG, et al. MicroRNA signatures associated with cytogenetics and prognosis in acute myeloid leukemia. Blood 2008; 111:3183.
  202. Jongen-Lavrencic M, Sun SM, Dijkstra MK, et al. MicroRNA expression profiling in relation to the genetic heterogeneity of acute myeloid leukemia. Blood 2008; 111:5078.
  203. Mi S, Lu J, Sun M, et al. MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia. Proc Natl Acad Sci U S A 2007; 104:19971.
  204. Li Z, Lu J, Sun M, et al. Distinct microRNA expression profiles in acute myeloid leukemia with common translocations. Proc Natl Acad Sci U S A 2008; 105:15535.
  205. Havelange V, Garzon R, Croce CM. MicroRNAs: new players in acute myeloid leukaemia. Br J Cancer 2009; 101:743.
  206. Garzon R, Heaphy CE, Havelange V, et al. MicroRNA 29b functions in acute myeloid leukemia. Blood 2009; 114:5331.
  207. Ramsingh G, Koboldt DC, Trissal M, et al. Complete characterization of the microRNAome in a patient with acute myeloid leukemia. Blood 2010; 116:5316.
  208. Marcucci G, Mrózek K, Radmacher MD, et al. The prognostic and functional role of microRNAs in acute myeloid leukemia. Blood 2011; 117:1121.
  209. Schwind S, Maharry K, Radmacher MD, et al. Prognostic significance of expression of a single microRNA, miR-181a, in cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol 2010; 28:5257.
  210. Marcucci G, Maharry KS, Metzeler KH, et al. Clinical role of microRNAs in cytogenetically normal acute myeloid leukemia: miR-155 upregulation independently identifies high-risk patients. J Clin Oncol 2013; 31:2086.
  211. Laupeze B, Amiot L, Drenou B, et al. High multidrug resistance protein activity in acute myeloid leukaemias is associated with poor response to chemotherapy and reduced patient survival. Br J Haematol 2002; 116:834.
  212. Legrand O, Simonin G, Perrot JY, et al. Pgp and MRP activities using calcein-AM are prognostic factors in adult acute myeloid leukemia patients. Blood 1998; 91:4480.
  213. Karászi E, Jakab K, Homolya L, et al. Calcein assay for multidrug resistance reliably predicts therapy response and survival rate in acute myeloid leukaemia. Br J Haematol 2001; 112:308.
  214. Schaich M, Soucek S, Thiede C, et al. MDR1 and MRP1 gene expression are independent predictors for treatment outcome in adult acute myeloid leukaemia. Br J Haematol 2005; 128:324.
  215. Schaich M, Ritter M, Illmer T, et al. Mutations in ras proto-oncogenes are associated with lower mdr1 gene expression in adult acute myeloid leukaemia. Br J Haematol 2001; 112:300.
  216. Mahadevan D, List AF. Targeting the multidrug resistance-1 transporter in AML: molecular regulation and therapeutic strategies. Blood 2004; 104:1940.
  217. Tsuji K, Motoji T, Sugawara I, et al. Significance of lung resistance-related protein in the clinical outcome of acute leukaemic patients with reference to P-glycoprotein. Br J Haematol 2000; 110:370.
  218. Barjesteh van Waalwijk van Doorn-Khosrovani S, Erpelinck C, van Putten WL, et al. High EVI1 expression predicts poor survival in acute myeloid leukemia: a study of 319 de novo AML patients. Blood 2003; 101:837.
  219. Gröschel S, Lugthart S, Schlenk RF, et al. High EVI1 expression predicts outcome in younger adult patients with acute myeloid leukemia and is associated with distinct cytogenetic abnormalities. J Clin Oncol 2010; 28:2101.
  220. Campos L, Oriol P, Sabido O, Guyotat D. Simultaneous expression of P-glycoprotein and BCL-2 in acute myeloid leukemia blast cells. Leuk Lymphoma 1997; 27:119.
  221. Adida C, Recher C, Raffoux E, et al. Expression and prognostic significance of survivin in de novo acute myeloid leukaemia. Br J Haematol 2000; 111:196.
  222. Kharas MG, Lengner CJ, Al-Shahrour F, et al. Musashi-2 regulates normal hematopoiesis and promotes aggressive myeloid leukemia. Nat Med 2010; 16:903.
  223. Kolitz JE, George SL, Marcucci G, et al. P-glycoprotein inhibition using valspodar (PSC-833) does not improve outcomes for patients younger than age 60 years with newly diagnosed acute myeloid leukemia: Cancer and Leukemia Group B study 19808. Blood 2010; 116:1413.
  224. Gröschel S, Schlenk RF, Engelmann J, et al. Deregulated expression of EVI1 defines a poor prognostic subset of MLL-rearranged acute myeloid leukemias: a study of the German-Austrian Acute Myeloid Leukemia Study Group and the Dutch-Belgian-Swiss HOVON/SAKK Cooperative Group. J Clin Oncol 2013; 31:95.
  225. Lugthart S, Figueroa ME, Bindels E, et al. Aberrant DNA hypermethylation signature in acute myeloid leukemia directed by EVI1. Blood 2011; 117:234.
  226. Brouwer RE, Hoefnagel J, Borger van Der Burg B, et al. Expression of co-stimulatory and adhesion molecules and chemokine or apoptosis receptors on acute myeloid leukaemia: high CD40 and CD11a expression correlates with poor prognosis. Br J Haematol 2001; 115:298.
  227. Ong YL, McMullin MF, Bailie KE, et al. High bax expression is a good prognostic indicator in acute myeloid leukaemia. Br J Haematol 2000; 111:182.
  228. Del Poeta G, Venditti A, Del Principe MI, et al. Amount of spontaneous apoptosis detected by Bax/Bcl-2 ratio predicts outcome in acute myeloid leukemia (AML). Blood 2003; 101:2125.
  229. Konopleva M, Pollyea DA, Potluri J, et al. Efficacy and Biological Correlates of Response in a Phase II Study of Venetoclax Monotherapy in Patients with Acute Myelogenous Leukemia. Cancer Discov 2016; 6:1106.
  230. Nakase K, Kita K, Otsuji A, et al. Diagnostic and clinical importance of interleukin-2 receptor alpha chain expression on non-T-cell acute leukaemia cells. Br J Haematol 1992; 80:317.
  231. Terwijn M, Feller N, van Rhenen A, et al. Interleukin-2 receptor alpha-chain (CD25) expression on leukaemic blasts is predictive for outcome and level of residual disease in AML. Eur J Cancer 2009; 45:1692.
  232. Gönen M, Sun Z, Figueroa ME, et al. CD25 expression status improves prognostic risk classification in AML independent of established biomarkers: ECOG phase 3 trial, E1900. Blood 2012; 120:2297.
Topic 4516 Version 69.0

References